Exosomes derived from MSCs ameliorate retinal laser injury partially by inhibition of MCP-1

Bo Yu, Hui Shao, Chang Su, Yuanfeng Jiang, Xiteng Chen, Lingling Bai, Yan Zhang, Qiutang Li, Xiaomin Zhang, Xiaorong Li, Bo Yu, Hui Shao, Chang Su, Yuanfeng Jiang, Xiteng Chen, Lingling Bai, Yan Zhang, Qiutang Li, Xiaomin Zhang, Xiaorong Li

Abstract

Although accumulated evidence supports the notion that mesenchymal stem cells (MSCs) act in a paracrine manner, the mechanisms are still not fully understood. Recently, MSC-derived exosomes (MSC-Exos), a type of microvesicle released from MSCs, were thought to carry functional proteins and RNAs to recipient cells and play therapeutic roles. In the present study, we intravitreally injected MSCs derived from either mouse adipose tissue or human umbilical cord, and their exosomes to observe and compare their functions in a mouse model of laser-induced retinal injury. We found that both MSCs and their exosomes reduced damage, inhibited apoptosis, and suppressed inflammatory responses to obtain better visual function to nearly the same extent in vivo. Obvious down-regulation of monocyte chemotactic protein (MCP)-1 in the retina was found after MSC-Exos injection. In vitro, MSC-Exos also down-regulated MCP-1 mRNA expression in primarily cultured retinal cells after thermal injury. It was further demonstrated that intravitreal injection of an MCP-1-neutralizing antibody promoted the recovery of retinal laser injury, whereas the therapeutic effect of exosomes was abolished when MSC-Exos and MCP-1 were administrated simultaneously. Collectively, these results suggest that MSC-Exos ameliorate laser-induced retinal injury partially through down-regulation of MCP-1.

Figures

Figure 1
Figure 1
Micrographs of scanning electron microscopy of (A) maMSC-Exos, (B) hucMSC-Exos and (C) f-Exos show spheroid shaped vesicles at the diameter of about 40–100 nm. Scale bar = 200 nm.
Figure 2. MaMSCs and maMSC-Exos promote the…
Figure 2. MaMSCs and maMSC-Exos promote the recovery of laser-induced retinal injury.
(A) Micrographs of retinal histological images (H&E staining) before injury (A1) and 3 days after injury (A2). Main histological parameters are presented with lines. Scale bar = 50 μm. (B) Effect of maMSC-Exos at different concentrations on diameter of retinal disordered area (B1) and ONL defect area (B2) at 3rd day post-injury. (C) Lesion areas after treatment with PBS, maMSCs or maMSC-Exos on 1st, 3rd, 7th and 14th days post-injury. n > 6, *p < 0.05. (D1) Amplitudes of dark-adapted ERG at 3, 14 and 60 days post-injury of PBS, maMSC- or maMSC-Exo-treated group. A wave and b wave amplitudes of dark adapted ERG (D2,D3) and light-adapted ERG (D4,D5) at 3, 7, 14, 21, 40 and 60 days post-injury were analysed and compared among the three groups. n = 7, *p < 0.05 (E) TUNEL staining of injury sections treated with PBS, maMSCs or maMSC-Exos at 1, 3, 7 and 14 days post-injury. n = 8, *p < 0.05. (F) Diameter of retinal disordered areas (F1), diameter of ONL defect areas (F2) and ERG responses (F3) at 3 days post-injury of maMSC-Exo- and f-Exo-treated groups. n = 6, *p < 0.05.
Figure 3. Representative examples of H&E staining…
Figure 3. Representative examples of H&E staining images at different time points of evaluation.
One day after injury of PBS (A1), maMSC-Exo (B1) or maMSC (C1) treated group. Three days after injury of PBS (A2), maMSC-Exo (B2) or maMSC (C2) treated group. Seven days after injury of PBS (A3), maMSC-Exo (B3) or maMSC (C3) treated group. Fourteen days after injury of PBS (A4), maMSC-Exo (B4) or maMSC (C4) treated group. Scale bar = 50 μm.
Figure 4. Representative examples of TUNEL staining…
Figure 4. Representative examples of TUNEL staining images at different time points of evaluation.
One day after injury of PBS(A1), maMSC-Exo (B1) or maMSC (C1) treated group. Three days after injury of PBS (A2), maMSC-Exo (B2) or maMSC (C2) treated group. Seven days after injury of PBS (A3), maMSC-Exo (B3) or maMSC (C3) treated group. Fourteen days after injury of PBS(A4), maMSC-Exo (B4) or maMSC (C4) treated group.(Scale bar = 20 μm; green: TUNEL staining of apoptotic cells; blue: DAPI staining of nucleus).
Figure 5. HucMSCs and hucMSC-Exos help protect…
Figure 5. HucMSCs and hucMSC-Exos help protect retina in mouse retinal laser injury model.
(A) Diameter of retinal disordered areas (A1) and ONL defect areas (A2) after treatment with PBS, hucMSCs or hucMSC-Exos at 1, 3, 7 and 14 days post-injury. n > 6, *p < 0.05. (B) Number of TUNEL positive cells/section of PBS-, hucMSC- and hucMSC-Exo-treated groups on 1st, 3rd, 7th and 14th days post-injury. n = 6, *p < 0.05. (C) A wave and b wave amplitudes of dark adapted ERG (C1,C2) and light-adapted ERG (C3,C5) at 3, 7, 14, 21, 40 and 60 days post-injury were analysed and compared among the three groups. n = 7, *p < 0.05.
Figure 6. MaMSC-Exos suppress the injury-induced inflammation…
Figure 6. MaMSC-Exos suppress the injury-induced inflammation in vivo and in vitro.
(A1–A3) Relative mRNA expression of MCP-1, TNF-α and ICAM1 of PBS- or MaMSC-Exo-treated group at 1, 3, 7 and 14 days post-injury. n = 7, *p B) Immunohistochemistry staining images of PBS- or MaMSC-Exo-treated group at 3 days post-injury. (C1,C2) Western blotting analyses of MCP-1 in retina/RPE/Choroid tissues from PBS- or MaMSC-Exo-treated group at 1, 3, 7 and 14 days post-injury. n = 3, *p < 0.05. (D1) Cell number of normal, thermo injury or MaMSC-Exo-treated group at 3 days after thermo injury. (D2) Relative mRNA expression of MCP-1 in normal, thermo injury or MaMSC-Exo-treated group at 3 days after thermo injury.
Figure 7. Representative examples of immunohistochemistry staining…
Figure 7. Representative examples of immunohistochemistry staining images at different time points of evaluation.
One day after injury of PBS (A1) or maMSC-Exo (B1) treated group. Seven days after injury of PBS (A1) or maMSC-Exo (B1) treated group. Fourteen days after injury of PBS (A1) or maMSC-Exo (B1) treated group. Scale bar = 20 μm.
Figure 8. Effect of MCP-1 regulation on…
Figure 8. Effect of MCP-1 regulation on laser induced retinal injury in mice.
(A1) Representative H&E staining images of PBS- or MCP-1 neutralizing antibody-treated groups at 3 days post-injury. Scale bar = 50 μm. Diameter of retinal disordered areas (A2) and diameter of ONL defect areas (A3) of PBS- or MCP-1 neutralizing antibody-treated group at 3 days post-injury. n = 6, *p 
All figures (8)

References

    1. Yoles E. & Schwartz M. Degeneration of spared axons following partial white matter lesion: implications for optic nerve neuropathies. Exp Neurol 153, 1–7 (1998).
    1. Zhou X., Xia X. B. & Xiong S. Q. Neuro-protection of retinal stem cells transplantation combined with copolymer-1 immunization in a rat model of glaucoma. Mol Cell Neurosci 54, 1–8 (2013).
    1. Gupta V. K., You Y., Li J. C., Klistorner A. & Graham S. L. Protective effects of 7,8-dihydroxyflavone on retinal ganglion and RGC-5 cells against excitotoxic and oxidative stress. J Mol Neurosci 49, 96–104 (2013).
    1. Gabelt B. et al.. Structure/function studies and the effects of memantine in monkeys with experimental glaucoma. Investig Ophthalmol Vis Sci 53, 2368–2376 (2012).
    1. Mayama C. Calcium channels and their blockers in intraocular pressure and glaucoma. Eur J Pharmacol 739, 96–105 (2013).
    1. Roh M. et al.. Etanercept, a widely used inhibitor of tumor necrosis factor-α (TNF-α), prevents retinal ganglion cell loss in a rat model of glaucoma. PLoS One 7, e40065 (2012).
    1. Kim B. et al.. In vitro and in vivo neuroprotective effects of cJun N-terminal kinase inhibitors on retinal ganglion cells. Mol Neurodegener 11, 30 (2016).
    1. Parrilla-Reverter G. et al.. Effects of different neurotrophic factors on the survival of retinal ganglion cells after a complete intraorbital nerve crush injury: a quantitative in vivo study. Exp Eye Res 89, 32–41 (2009).
    1. Afarid M., Torabi-Nami M. & Zare B. Neuroprotective and restorative effects of the brain-derived neurotrophic factor in retinal diseases. J Neurol Sci 363, 43–50 (2016).
    1. Matsuse D. et al.. Human umbilical cord-derived mesenchymal stromal cells differentiate into functional Schwann cells that sustain peripheral nerve regeneration. J Neuropathol Exp Neurol 69, 973–985 (2010).
    1. Reinshagen H. et al.. Corneal surface reconstruction using adult mesenchymal stem cells in experimental limbal stem cell deficiency in rabbits. Acta Ophthalmol 89, 741–748 (2011).
    1. Zhang X. et al.. Mesenchymal stem cells ameliorate experimental autoimmune uveoretinitis by comprehensive modulation of systemic autoimmunity. Invest Ophthalmol Vis Sci 52, 3143–3152 (2011).
    1. Machalinska A. et al.. Long-term neuroprotective effects of NT-4-engineered mesenchymal stem cells injected intravitreally in a mouse model of acute retinal injury. Invest Ophthalmol Vis Sci 54, 8292–8305 (2013).
    1. Rotenstreich Y. et al.. [A novel system for minimally invasive transplantation of bone marrow derived stem cells as a thin layer in the subretina and extravascular spaces of the choroid--for treatment of retinal degeneration. Harefuah 154, 84–88, 138 (2015).
    1. Zhang L. et al.. Long-term therapeutic effects of mesenchymal stem cells compared to dexamethasone on recurrent experimental autoimmune uveitis of rats. Invest Ophthalmol Vis Sci 55, 5561–5571 (2014).
    1. Timmers L. et al.. Reduction of myocardial infarct size by human mesenchymal stem cell conditioned medium. Stem Cell Res 1, 129–137 (2007).
    1. Bonventre J. V. Microvesicles from mesenchymal stromal cells protect against acute kidney injury. J Am Soc Nephrol 20, 927–928 (2009).
    1. Schorey J. S. & Bhatnagar S. Exosome function: from tumor immunology to pathogen biology. Traffic 9, 871–881 (2008).
    1. Yu B., Zhang X. & Li X. Exosomes derived from mesenchymal stem cells. Int J Mol Sci 15, 4142–4157 (2014).
    1. Herberts C. A., Kwa M. S. & Hermsen H. P. Risk factors in the development of stem cell therapy. J Transl Med 9, 29 (2011).
    1. Chen K. et al.. False-positive immunogenicity responses are caused by CD20+ B cell membrane fragments in an anti-ofatumumab antibody bridging assay. J Immunol Methods 394, 22–31 (2013).
    1. Xin H., Li Y. & Chopp M. Exosomes/miRNAs as mediating cell-based therapy of stroke. Front Cell Neurosci 8, 377 (2014).
    1. Anthony D. F. & Shiels P. G. Exploiting paracrine mechanisms of tissue regeneration to repair damaged organs. Transplant Res 2, 10 (2013).
    1. Jiang Y. et al.. Therapeutic effect of bone marrow mesenchymal stem cells on laser-induced retinal injury in mice. Int J Mol Sci 15, 9372–9385 (2014).
    1. Solberg Y., Rosner M., Turetz J. & Belkin M. MK-801 has neuroprotective and antiproliferative effects in retinal laser injury. Invest Ophthalmol Vis Sci 38, 1380–1389 (1997).
    1. Nonaka A. et al.. Inflammatory response after scatter laser photocoagulation in nonphotocoagulated retina. Invest Ophthalmol Vis Sci 43, 1204–1209 (2002).
    1. Zeng H. Y. et al.. Identification of sequential events and factors associated with microglial activation, migration, and cytotoxicity in retinal degeneration in rd mice. Invest Ophthalmol Vis Sci 46, 2992–2999 (2005).
    1. Lai R. C. et al.. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res 4, 214–222 (2010).
    1. Reis L. A. et al.. Bone marrow-derived mesenchymal stem cells repaired but did not prevent gentamicin-induced acute kidney injury through paracrine effects in rats. PLoS One 7, e44092 (2012).
    1. Zhang B. et al.. Mesenchymal stem cells secrete immunologically active exosomes. Stem Cells Dev 23, 1233–1244 (2014).
    1. Zhang B. et al.. HucMSC-Exosome Mediated-Wnt4 Signaling Is Required for Cutaneous Wound Healing. Stem Cells (2014).
    1. Xin H. et al.. Systemic administration of exosomes released from mesenchymal stromal cells promote functional recovery and neurovascular plasticity after stroke in rats. J Cereb Blood Flow Metab 33, 1711–1715 (2013).
    1. Belokopytov M., Shulman S., Dubinsky G., Belkin M. & Rosner M. Intravitreal saline injection ameliorates laser-induced retinal damage in rats. Retina 32, 1165–1170 (2012).
    1. Shulman S., Belokopytov M., Dubinsky G., Belkin M. & Rosner M. Ameliorative effect of PN-277 on laser-induced retinal damage. Graefes Arch Clin Exp Ophthalmol 247, 343–348 (2009).
    1. Ben-Shlomo G. et al.. Functional deficits resulting from laser-induced damage in the rat retina. Lasers Surg Med 38, 689–694 (2006).
    1. Sokolova V. et al.. Characterisation of exosomes derived from human cells by nanoparticle tracking analysis and scanning electron microscopy. Colloids Surf B Biointerfaces 87, 146–150 (2011).
    1. Ghazi-Nouri S. M. et al.. Laser photocoagulation alters the pattern of staining for neurotrophin-4, GFAP, and CD68 in human retina. Br J Ophthalmol 87, 488–492 (2003).
    1. Ng T. F., Turpie B. & Masli S. Thrombospondin-1-mediated regulation of microglia activation after retinal injury. Invest Ophthalmol Vis Sci 50, 5472–5478 (2009).
    1. Ajuebor M. N. et al.. Endogenous monocyte chemoattractant protein-1 recruits monocytes in the zymosan peritonitis model. J Leukoc Biol 63, 108–116 (1998).
    1. Yoshimura T., Robinson E. A., Tanaka S., Appella E. & Leonard E. J. Purification and amino acid analysis of two human monocyte chemoattractants produced by phytohemagglutinin-stimulated human blood mononuclear leukocytes. J Immunol 142, 1956–1962 (1989).
    1. Nakazawa T. et al.. Monocyte chemoattractant protein 1 mediates retinal detachment-induced photoreceptor apoptosis. Proc Natl Acad Sci USA 104, 2425–2430 (2007).
    1. Levy O. et al.. APOE Isoforms Control Pathogenic Subretinal Inflammation in Age-Related Macular Degeneration. J Neurosci 35, 13568–13576 (2015).
    1. Nawaz M. I. et al.. Autocrine CCL2, CXCL4, CXCL9 and CXCL10 signal in retinal endothelial cells and are enhanced in diabetic retinopathy. Exp Eye Res 109, 67–76 (2013).
    1. El-Asrar A. M. et al.. CXC chemokine expression profiles in aqueous humor of patients with different clinical entities of endogenous uveitis. Immunobiology 216, 1004–1009 (2011).
    1. Harada T. et al.. Microglia-Muller glia cell interactions control neurotrophic factor production during light-induced retinal degeneration. J Neurosci 22, 9228–9236 (2002).
    1. Dong N. et al.. Upregulation of retinal neuronal MCP-1 in the rodent model of diabetic retinopathy and its function in vitro. Invest Ophthalmol Vis Sci 53, 7567–7575 (2012).
    1. Raoul W. et al.. CCL2/CCR2 and CX3CL1/CX3CR1 chemokine axes and their possible involvement in age-related macular degeneration. J Neuroinflammation 7, 87 (2010).
    1. Sennlaub F. et al.. CCR2(+) monocytes infiltrate atrophic lesions in age-related macular disease and mediate photoreceptor degeneration in experimental subretinal inflammation in Cx3cr1 deficient mice. EMBO Mol Med 5, 1775–1793 (2013).
    1. Vlassov A. V., Magdaleno S., Setterquist R. & Conrad R. Exosomes: current knowledge of their composition, biological functions, and diagnostic and therapeutic potentials. Biochim Biophys Acta 1820, 940–948 (2012).
    1. Xin H. et al.. Exosome-mediated transfer of miR-133b from multipotent mesenchymal stromal cells to neural cells contributes to neurite outgrowth. Stem Cells 30, 1556–1564 (2012).
    1. Koh W. et al.. Analysis of deep sequencing microRNA expression profile from human embryonic stem cells derived mesenchymal stem cells reveals possible role of let-7 microRNA family in downstream targeting of hepatic nuclear factor 4 alpha. BMC Genomics 11 Suppl 1, S6 (2010).
    1. Lai R. C., Chen T. S. & Lim S. K. Mesenchymal stem cell exosome: a novel stem cell-based therapy for cardiovascular disease. Regen Med 6, 481–492 (2011).

Source: PubMed

3
Prenumerera